KEGG: nfi:NFIA_052450
STRING: 36630.CADNFIAP00004851
Recombinant Neosartorya fischeri Probable carboxypeptidase NFIA_052450 is a protein from the fungus Neosartorya fischeri (also known as Aspergillus fischeri) produced through recombinant DNA technology, typically in E. coli expression systems . It belongs to the carboxypeptidase family of enzymes, which perform diverse physiological functions by removing C-terminal amino acids from proteins and peptides .
The significance of this protein in research stems from several areas:
As a member of the metallocarboxypeptidase family, it may have roles in protein processing and modification
Potential applications in understanding fungal physiology and metabolism
Possible relevance to the development of antifungal strategies, given that other proteins from N. fischeri have demonstrated antifungal properties
Unlike the well-characterized antifungal proteins NFAP and NFAP2 from the same organism, NFIA_052450 remains less studied but represents an important target for enzyme research and potential biotechnological applications.
Carboxypeptidases in fungi like Neosartorya fischeri perform several critical physiological functions:
Protein Processing: They participate in post-translational modification by removing C-terminal amino acids from proteins and peptides .
Nutrient Acquisition: In fungi, these enzymes can facilitate the breakdown of extracellular proteins into assimilable nitrogen sources.
Cellular Metabolism: They may be involved in the regulation of protein turnover and homeostasis.
Specialized Functions: Some fungal carboxypeptidases have evolved specialized roles, such as participating in the production of bioactive compounds .
The diversity of carboxypeptidases in fungi suggests their adaptation to various cellular processes and environmental conditions. In N. fischeri specifically, other enzymes such as β-glucosidase (NfBGL1) have been characterized with roles in substrate hydrolysis , indicating a diverse enzymatic repertoire in this organism that likely includes specialized carboxypeptidases.
Expression and purification of Recombinant NFIA_052450 can be approached using methodologies demonstrated effective for other Neosartorya fischeri proteins:
Expression Systems:
Pichia pastoris: Successfully used for other N. fischeri proteins like NFAP
Penicillium chrysogenum-based system: Shown to be effective for NFAP2 expression
Recommended Expression Protocol:
Clone the NFIA_052450 gene into an appropriate expression vector
Transform the vector into the chosen expression host
Induce protein expression (e.g., with IPTG for E. coli or methanol for P. pastoris)
Harvest cells and extract the recombinant protein
Purification Strategy:
Initial clarification of cell lysate
Affinity chromatography (if the protein is tagged)
Ion-exchange chromatography for further purification
Size-exclusion chromatography as a polishing step
Heat treatment may be applicable if the protein is thermostable, as observed with other N. fischeri proteins
Verification Methods:
SDS-PAGE to confirm protein size
Western blotting for identity confirmation
Mass spectrometry for accurate molecular weight determination
Activity assays to confirm enzymatic function
For optimal results, consider including a purification tag (His-tag, GST, etc.) in the recombinant construct to facilitate affinity purification, with an option for tag removal if needed for functional studies.
Characterizing the enzymatic activity of recombinant NFIA_052450 requires targeted approaches based on its predicted carboxypeptidase function:
Substrate Selection:
Synthetic peptides with C-terminal modifications
Fluorogenic or chromogenic substrates for continuous monitoring
Natural protein substrates to assess physiological relevance
Activity Assay Methods:
Spectrophotometric assays: Monitor the release of amino acids with specific reagents
Fluorometric assays: Using fluorescence-quenched substrates
HPLC-based assays: For precise quantification of cleaved products
Mass spectrometry: For detailed substrate specificity analysis
Optimal Assay Conditions Determination:
pH optimization (typically test range pH 4.0-9.0)
Temperature profiling (25-80°C based on N. fischeri enzyme thermostability)
Metal ion requirements (test Zn²⁺, Ca²⁺, Mg²⁺, Mn²⁺)
Buffer composition effects
Kinetic Parameter Determination:
Employ Multiple Injection Method (MIM) with Isothermal Titration Calorimetry (ITC) as described for enzyme kinetics :
Parameter Measurement | Experimental Approach | Analysis Method |
---|---|---|
KM and Vmax | Substrate concentration series | Michaelis-Menten kinetics |
kcat | Enzyme concentration variation | Linear regression |
Inhibition constants | Inhibitor titration | Dixon plots or IC50 determination |
For optimal experimental design, implement the penalized expectation of determinant (ED)-optimal design approach to reduce the uncertainty of parameter estimates , which has been shown to improve the Cramer-Rao lower bound of the variance of parameter estimation error to 82% for μmax and to 60% for KM compared to batch experiments.
While specific structural and functional data for NFIA_052450 is limited, we can draw comparisons with other characterized proteins from Neosartorya fischeri:
Structural Comparison:
Functional Comparison:
NFIA_052450: Probable carboxypeptidase activity, specific substrates unknown
NFAP: Antifungal activity against filamentous fungi, disrupts cell wall organization
NFAP2: Potent anti-Candida activity, potential clinical applications
NfBGL1: β-glucosidase activity, converts isoflavone glycosides to aglycones
Evolutionary Context:
The diverse functional profiles of proteins from N. fischeri suggest adaptation to various ecological niches. While NFAP and NFAP2 appear specialized for antimicrobial defense, NFIA_052450 likely serves a different physiological role, potentially in metabolism or protein processing.
Unlike the well-characterized antifungal proteins, NFIA_052450's specific function remains to be elucidated through detailed biochemical characterization, but methods deployed for other N. fischeri proteins provide a roadmap for its investigation.
Researchers expressing NFIA_052450 heterologously may encounter several challenges based on experiences with other Neosartorya fischeri proteins:
Expression Yield Limitations:
Low-yield production has been reported as a limiting factor for native N. fischeri proteins
The average yield of recombinant NFAP2 was 40-times higher than in the native producer, indicating optimization is essential
Protein Folding and Processing:
Correct disulfide bond formation may be critical if NFIA_052450 contains multiple cysteine residues (as seen with NFAP)
Post-translational modifications present in the native protein may be absent in prokaryotic expression systems
Challenges by Expression System:
Purification Challenges:
Proteolytic degradation during extraction
Co-purification of host cell proteins with similar properties
Maintaining enzyme activity during purification steps
Solutions from Literature:
Studies with NFAP and NFAP2 have overcome these challenges through:
Use of fusion tags to enhance solubility and facilitate purification
High-cell-density fermentation techniques yielding up to 1873±1.5 U/ml activity
Development of alternative production methods such as synthetic peptide synthesis and native chemical ligation
A systematic approach to expression optimization is recommended, testing multiple expression constructs, hosts, and conditions in parallel.
Determining kinetic parameters of NFIA_052450 requires sophisticated experimental design to ensure accurate and reliable results:
Optimal Experimental Design Strategy:
Preliminary Experimental Planning:
Employ penalized expectation of determinant (ED)-optimal design with discrete parameter distribution
Optimize sample times and starting concentrations (C₀) to minimize uncertainty of parameter estimates
For substrate feed-batch process design, implement small volume flow which has been shown to reduce parameter estimation error compared to batch experiments
Calorimetric Approach:
Data Collection Parameters:
Parameter | Recommended Range | Optimization Consideration |
---|---|---|
Substrate concentrations | 0.01-100 μM | Cover 0.1×KM to 10×KM |
Enzyme concentrations | Picomolar to nanomolar | Ensure <5% substrate depletion between injections |
Temperature | 25-37°C | Physiologically relevant |
Injection volumes | 1-3 μL (low volume) or 3-8 μL (standard) | Optimize signal-to-noise ratio |
Time between injections | 2-4 minutes | Allow steady state establishment |
Data Analysis Framework:
Plot reaction rate (d[P]/dt) versus substrate concentration [S]
Fit to Michaelis-Menten equation: v = (Vmax × [S])/(KM + [S])
Analyze with Lineweaver-Burk double reciprocal plot for visualization of inhibition patterns
Calculate kcat from Vmax/[E]total
Validation Approaches:
Perform experiments at multiple enzyme concentrations
Compare results from different methodologies (spectrophotometric, fluorometric)
Assess impact of potential inhibitors and activators
This approach has been shown to reduce the Cramer-Rao lower bound of the variance of parameter estimation error to 82% for μmax and to 60% for KM compared to conventional batch experiments .
Investigating the substrate specificity of NFIA_052450 requires a multi-faceted approach to understand both the range of acceptable substrates and the structural determinants of specificity:
Comprehensive Substrate Profiling:
Peptide Library Screening:
Utilize positional scanning synthetic combinatorial libraries
Test peptides with varying C-terminal residues to determine P1' preferences
Analyze upstream residue preferences (P1, P2, P3) using systematically varied peptides
Carboxypeptidase B-Assisted Charge-Based Fractional Diagonal Chromatography (CPB-ChaFRADIC):
Mass Spectrometry-Based Approaches:
Perform LC-MS/MS analysis of digestion products
Identify cleavage sites through comparative peptide mapping
Quantify relative cleavage efficiencies with isotope-labeled substrates
Structural Basis of Specificity:
Homology Modeling and Docking Studies:
Generate structural models based on related carboxypeptidases
Identify putative substrate binding pockets and catalytic residues
Perform in silico docking to predict substrate interactions
Site-Directed Mutagenesis:
Mutate predicted substrate-binding residues
Analyze shifts in specificity or activity following mutations
Create a structure-function map of the substrate binding pocket
Experimental Validation Framework:
Substrate Type | Analytical Method | Expected Outcome |
---|---|---|
Synthetic peptides | HPLC, MS | Identification of preferred C-terminal residues |
Natural proteins | SDS-PAGE, MS | Physiologically relevant substrates |
Inhibitors | Enzyme kinetics | Binding pocket characteristics |
Fluorogenic substrates | Fluorescence spectroscopy | Real-time activity monitoring |
Based on observations with carboxypeptidase B, which selectively catalyzes the release of C-terminal lysine and arginine , particular attention should be paid to basic amino acids in the substrate screening, while recognizing that NFIA_052450 may have different specificity.
For comprehensive analysis, include examination of:
pH dependence of substrate preferences
Metal ion effects on specificity
Temperature influence on substrate selection
Competitive substrate assays to determine relative preferences
Understanding the biological role of NFIA_052450 in Neosartorya fischeri requires integrative approaches spanning genetics, biochemistry, and systems biology:
Hypothesized Biological Functions:
Based on carboxypeptidase roles in other systems and the biology of N. fischeri, NFIA_052450 might function in:
Nutrient acquisition through protein degradation
Cell wall remodeling during growth and development
Post-translational processing of secreted proteins
Detoxification of harmful peptides in the environment
Regulation of signaling peptides
Investigative Approaches:
Genetic Manipulation Studies:
Biochemical and Proteomic Approaches:
Identify natural substrates through pull-down experiments
Characterize the secretome and peptidome in wild-type vs. NFIA_052450 mutants
Analyze changes in cell wall composition in mutants
Perform metabolomic profiling to identify pathways affected by NFIA_052450 activity
Localization and Expression Studies:
Fluorescent protein tagging to determine subcellular localization
Immunolocalization with specific antibodies
qRT-PCR to measure expression under various conditions
RNA-seq to identify co-regulated genes
Environmental Response Characterization:
Environmental Condition | Measurement | Potential Insight |
---|---|---|
Nutrient limitation | Expression changes | Role in nutrient acquisition |
Cell wall stress | Growth, morphology | Involvement in cell wall integrity |
pH variation | Activity, expression | Environmental adaptation function |
Presence of competitors | Secretion patterns | Role in competitive interactions |
Developmental stages | Temporal expression | Function in lifecycle progression |
Comparative Genomics:
Analyze conservation of NFIA_052450 across fungal species
Identify syntenic relationships and gene neighborhoods
Examine evolutionary patterns suggestive of functional constraints
Compare with carboxypeptidases of known function in other organisms
For N. fischeri specifically, the thermal stability of its enzymes (with temperature optima up to 80°C for some enzymes ) suggests investigating how NFIA_052450 might contribute to thermoadaptation and survival in high-temperature environments.
Site-directed mutagenesis offers a powerful approach to dissect the structure-function relationships in NFIA_052450, providing insights into catalytic mechanism, substrate specificity, and protein stability:
Strategic Mutagenesis Targets:
Catalytic Residues:
Identify putative catalytic triad/dyad based on homology to characterized carboxypeptidases
Create conservative mutations (e.g., Glu→Asp, His→Asn) to assess catalytic requirements
Generate complete knockout mutations to confirm essentiality
Metal-Binding Sites:
If NFIA_052450 is a metallocarboxypeptidase, mutate predicted metal-coordinating residues
Analyze effects on activity, stability, and metal preference
Create variants with altered metal specificity
Substrate-Binding Pocket:
Target residues lining the predicted S1' pocket that interacts with the C-terminal residue
Introduce mutations that alter pocket size, hydrophobicity, and charge
Engineer specificity shifts by rational design of binding site residues
Structural Elements:
Mutagenesis Design Framework:
Mutation Type | Design Strategy | Expected Outcome | Analysis Method |
---|---|---|---|
Alanine scanning | Replace key residues with alanine | Identify essential residues | Activity assays |
Conservative substitutions | Replace with similar amino acids | Role of specific properties | Kinetic analysis |
Non-conservative substitutions | Replace with dissimilar amino acids | Engineering new functions | Substrate profiling |
Deletion mutants | Remove structural elements | Importance of domains | Structural analysis |
Charge reversal | Replace basic with acidic or vice versa | Electrostatic contributions | pH profiles |
Integrated Analysis Approach:
Functional Characterization:
Compare enzyme kinetics (kcat, KM) between wild-type and mutants
Assess substrate specificity changes using peptide libraries
Determine pH-activity and temperature-activity profiles
Structural Verification:
Computational Support:
Molecular dynamics simulations to predict effects of mutations
Homology modeling to visualize structural changes
Docking studies to predict altered substrate interactions
This comprehensive mutagenesis approach, similar to that applied to NFAP , will reveal which structural elements are critical for NFIA_052450's folding, stability, and catalytic function, providing a foundation for potential protein engineering applications.
Investigating applications of NFIA_052450 in biotechnology and medicine requires systematic exploration of its properties and potential uses based on its enzymatic activity:
Biotechnological Applications Assessment:
Biocatalysis Applications:
Screen activity on industrially relevant substrates
Evaluate performance under process-relevant conditions (temperature, pH, solvents)
Assess stability during prolonged reactions and storage
Compare efficiency to commercially used enzymes
Food and Beverage Processing:
Test protein modification capabilities in food matrices
Evaluate flavor enhancement potential through peptide modification
Assess allergen reduction capabilities through specific peptide cleavage
Investigate texture modification properties
Agricultural Applications:
Medical Applications Exploration:
Therapeutic Potential Assessment:
Investigate selective proteolytic capabilities for:
Bioactive peptide production
Protein drug modification
Diagnostic applications
Screen for inhibition of pathologically relevant peptides
Anti-Fungal Applications:
Safety and Production Considerations:
Assessment Area | Methods | Critical Parameters |
---|---|---|
Cytotoxicity | Human cell line assays | Cell viability, membrane integrity |
Immunogenicity | In silico epitope prediction, blood cell assays | Inflammatory markers, antibody production |
Stability | Accelerated stability testing | Half-life, activity retention |
Scale-up potential | Pilot-scale production trials | Yield, purification efficiency, cost |
Engineering for Application:
Develop immobilization strategies for reuse and stability enhancement
Engineer variants with improved properties (thermostability, pH tolerance)
Design fusion proteins for targeted applications
Optimize formulation for specific delivery methods
Drawing from the experience with NFAP and NFAP2, which demonstrated both agricultural and clinical potential , a parallel exploration of NFIA_052450's capabilities could reveal unique applications based on its carboxypeptidase activity.
Determining the three-dimensional structure of NFIA_052450 requires sophisticated structural biology techniques. Based on approaches used for other Neosartorya fischeri proteins, the following methods are recommended:
X-ray Crystallography Approach:
Crystallization Screening:
Implement sparse matrix screening with commercial kits
Explore crystallization with bound substrates or inhibitors
Test seeding techniques to improve crystal quality
Consider surface entropy reduction mutations to enhance crystallizability
Data Collection and Processing:
Collect high-resolution diffraction data at synchrotron facilities
Process data with modern software packages (XDS, DIALS)
Consider experimental phasing using heavy atoms if molecular replacement fails
Implement anisotropic diffraction correction if necessary
Nuclear Magnetic Resonance (NMR) Spectroscopy:
Sample Preparation:
Produce isotopically labeled protein (¹⁵N, ¹³C, ²H)
Optimize buffer conditions for stability and spectral quality
Determine optimal temperature for data collection
Spectral Assignment and Structure Calculation:
Collect suite of 2D and 3D heteronuclear experiments
Assign backbone and side-chain resonances
Measure distance restraints using NOE experiments
Calculate structural ensemble using restrained molecular dynamics
Complementary Techniques:
Integration of Structural and Functional Data:
From studies with NFAP, we know that disulfide bridges, hydrophobic core, and N-terminal amino acids play crucial roles in forming stable, folded, and functionally active proteins from N. fischeri . Similar structural elements should be carefully characterized in NFIA_052450 to understand their contribution to carboxypeptidase activity.
Understanding the stability and activity profile of NFIA_052450 across various environmental conditions is crucial for both basic research and applications. Based on studies of other Neosartorya fischeri enzymes, a comprehensive characterization would include:
Temperature Effects:
Thermal Stability Analysis:
Determine melting temperature (Tm) using differential scanning calorimetry
Assess activity retention after heat treatment at various temperatures
Measure half-life at elevated temperatures
Compare with other N. fischeri enzymes, which have shown remarkable thermostability (e.g., NfBGL1 with 80°C temperature optimum)
Temperature-Activity Relationship:
Determine temperature optimum for enzymatic activity
Construct Arrhenius plot to calculate activation energy
Identify cold-activity properties if present
pH and Ionic Effects:
pH Stability Profile:
pH-Activity Relationship:
Determine optimal pH for enzymatic activity
Analyze pH-dependent changes in kinetic parameters
Identify ionizable groups involved in catalysis through pH-rate profiles
Ionic Strength Effects:
Solvent and Chemical Stability:
Organic Solvent Tolerance:
Test activity retention in water-miscible organic solvents
Determine concentration thresholds for activity loss
Evaluate potential for biocatalysis in non-aqueous media
Chemical Denaturant Resistance:
Environmental Stability Matrix:
Condition | Measurement Parameters | Expected Insights |
---|---|---|
Temperature | 4-90°C in 10°C increments | Thermostability profile |
pH | pH 2-12 in 1 unit increments | pH stability range |
Salt concentration | 0-2M NaCl, KCl, MgCl₂ | Ionic strength effects |
Storage stability | 4°C, -20°C, -80°C; 1-12 months | Long-term storage conditions |
Freeze-thaw cycles | 1-10 cycles | Process stability |
Metal ions | EDTA treatment, metal supplementation | Metal dependency |